Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Med Sci Monit ; 24: 2508-2517, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29687789

RESUMEN

BACKGROUND Myosin phosphatase target subunit 1 (MYPT1) serves as a subgroup of myosin phosphatases, and is frequently low-expressed in human cancers. However, little is known about the effects of MYPT1 in gastric cancer (GC). MATERIAL AND METHODS In our study, MYPT1 expression was detected by quantitative real-time reverse transcription PCR (qRT-PCR) in GC tissues, different advanced pathological stages of GC tissues, and preoperative and postoperative patients. Kaplan-Meier analysis was used to measure the overall survival of GC patients. MYPT1 expression was analyzed by qRT-PCR and Western blot assays in GES-1 cells and GC cells. Cell proliferation, cycle, and migration and invasion abilities were detected by CCK-8, flow cytometry, and Transwell assays. E-cadherin, TIMP-2, MMP-2, MMP-9 RhoA, and p-RhoA expressions were assessed by qRT-PCR and Western blot assays in treated SNU-5 cells. RESULTS Our results indicated that MYPT1 was down-regulated in GC tissues and cells, and is related to clinical stages and overall survival of GC. Functional research demonstrated that overexpression of MYPT1 can inhibit cell proliferation, cell cycle progression, and migration and invasion of GC cells. Many studies on mechanisms reported that overexpression of MYPT1 dramatically improved the expression levels of cell cycle-related genes (Cyclin D1 and c-myc), significantly increased epithelial marker (E-cadherin) expression, and decreased invasion-associated genes (TIMP-2 and MMP-2) expressions in SNU-5 cells. In addition, we found that MYPT1 suppressed RhoA phosphorylation. CONCLUSIONS We verified that MYPT1 inhibits GC cell proliferation and metastasis by regulating RhoA phosphorylation.


Asunto(s)
Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Neoplasias Gástricas/enzimología , Antígenos CD , Cadherinas/biosíntesis , Ciclo Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Pronóstico , Neoplasias Gástricas/sangre , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Factores de Transcripción/biosíntesis
2.
PLoS Genet ; 12(11): e1006440, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27855159

RESUMEN

During embryogenesis the spinal cord shifts position along the anterior-posterior axis relative to adjacent tissues. How motor neurons whose cell bodies are located in the spinal cord while their axons reside in adjacent tissues compensate for such tissue shift is not well understood. Using live cell imaging in zebrafish, we show that as motor axons exit from the spinal cord and extend through extracellular matrix produced by adjacent notochord cells, these cells shift several cell diameters caudally. Despite this pronounced shift, individual motoneuron cell bodies stay aligned with their extending axons. We find that this alignment requires myosin phosphatase activity within motoneurons, and that mutations in the myosin phosphatase subunit mypt1 increase myosin phosphorylation causing a displacement between motoneuron cell bodies and their axons. Thus, we demonstrate that spinal motoneurons fine-tune their position during axonogenesis and we identify the myosin II regulatory network as a key regulator.


Asunto(s)
Axones , Desarrollo Embrionario/genética , Fosfatasa de Miosina de Cadena Ligera/genética , Neurogénesis/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células Musculares/citología , Células Musculares/metabolismo , Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Notocorda/crecimiento & desarrollo , Médula Espinal/crecimiento & desarrollo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
3.
Oncol Rep ; 32(6): 2795-802, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25333508

RESUMEN

Inhibition of angiogenesis is an important therapeutic strategy for advanced stage prostate cancer (PCa). RhoA/Rho-associated protein kinases (ROCK) are key regulators of the cytoskeleton and have been implicated in PCa angiogenesis. We investigated the anti-angiogenic effects of fasudil, a ROCK inhibitor, on PCa-induced angiogenesis in vitro. Proliferation of PCa-conditioned human umbilical vein endothelial cells (HUVECs) was assessed using a bromodeoxyuridine (BrdU) assay, and migration was assessed with a wound healing assay. In vitro angiogenesis of PCa-conditioned HUVECs was evaluated by tube formation and a spheroid sprouting assay. Fasudil inhibited PCa-induced endothelial cell proliferation at a concentration of 100 µM, and also decreased PCa-induced endothelial cell migration at a concentration of 30 µM. In the in vitro angiogenesis assay, fasudil exerted a more significant effect. Tube formation was significantly inhibited at fasudil concentrations exceeding 3 µM, and spheroid sprouts were significantly thinner and shorter (at fasudil concentrations of 10 and 30 µM, respectively). Western blotting results showed that expression of phosphorylated myosin phosphatase target subunit 1 (MYPT-1) was significantly lower after fasudil treatment, confirming that fasudil inhibited ROCK activity in these model systems. These data suggest that fasudil may be a useful anti-angiogenic agent for PCa.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Quinasas Asociadas a rho/genética , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinasas Asociadas a rho/antagonistas & inhibidores
4.
Protein Expr Purif ; 81(1): 126-135, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22005452

RESUMEN

Protein-protein interactions between MBS and PKG are mediated by the involvement of C-terminal domain of MBS, MBS(CT180) and N-terminal coiled coil (CC) leucine zipper (LZ) domain of PKG-Iα, PKG-Iα1(-59). MBS(CT180) is comprised of three structurally variant domains of non-CC, CC, and LZ nature. Paucity of three-dimensional structural information of these MBS domains precludes atomic level understanding of MBS-PKG contractile complex structure. Here we present data on cloning, expression, and purification of CC, LZ, and CCLZ domains of MBS(CT180) and their biophysical characterization using size exclusion chromatography (SEC), circular dichroism (CD), and two-dimensional (1)H-(15)N HSQC NMR. The methods as detailed resulted in high level protein expression and high milligram quantities of purified isotopically ((15)N and (13)C) enriched polypeptides. SEC, CD, and (1)H-(15)N HSQC NMR experiments demonstrated that recombinantly expressed MBS CC domain is well folded and exists as a dimer within physiologic pH range, which is supported by our previous findings. The dimerization of CC MBS is likely mediated through formation of coiled coil conformation. In contrast, MBS LZ domain was almost unfolded that exists as non-stable low structured monomer within physiologic pH range. Protein folding and stability of MBS LZ was improved as a function of decrease in pH that adopts a folded, stable, and structured conformation at acidified pH 4.5. SEC and NMR analyses of LZ vs. CCLZ MBS domains indicated that inclusion of CC domain partially improves protein folding of LZ domain.


Asunto(s)
Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Proteínas Recombinantes/biosíntesis , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Cromatografía de Afinidad , Cromatografía en Gel , Dicroismo Circular , Clonación Molecular , Datos de Secuencia Molecular , Fosfatasa de Miosina de Cadena Ligera/química , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
5.
Biol Reprod ; 75(3): 452-61, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16554414

RESUMEN

RHO GTPases are key regulators of the actin cytoskeleton and stress fiber formation. In the human uterus, activated RHOA forms a complex with RHO-associated protein kinase (ROCK) which inhibits myosin light chain phosphatase (PPP1R12A), causing a calcium-independent increase in myosin light chain phosphorylation and tension (Ca2+ sensitization). Recently discovered small GTP binding RND proteins can inhibit RHOA and ROCK interaction to reduce calcium sensitization. Very little is known about the expression of RND proteins in the human uterus. We tested the hypothesis that the uterine quiescence observed during gestation is mediated by an increase in RND protein expression inhibiting RHOA-ROCK-mediated PPP1R12A phosphorylation. Immunohistochemistry and immunoblotting were used to determine RHOA and RND protein expression and localization in nonpregnant, pregnant nonlaboring, and laboring patients at term and patients in spontaneous preterm labor. Changes in protein expression estimated by densitometry between different patient groups were measured. A significant increase of RND2 and RND3 protein expression was observed in pregnant relative to nonpregnant myometrium associated with a loss of PPP1R12A phosphorylation. RND transfected myometrial cells demonstrated a dramatic loss of stress fiber formation and a "rounding" phenotype. RND upregulation in pregnancy may inhibit RHOA-ROCK-mediated increase in calcium sensitization to facilitate the uterine quiescence observed during gestation.


Asunto(s)
Miometrio/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Adulto , Western Blotting , Densitometría , Femenino , Técnica del Anticuerpo Fluorescente , GTP Fosfohidrolasas/biosíntesis , Humanos , Inmunohistoquímica , Técnicas In Vitro , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/genética , Persona de Mediana Edad , Músculo Liso/fisiología , Miometrio/citología , Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Fosfatasa de Miosina de Cadena Ligera/genética , Embarazo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Transfección , Útero/fisiología , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho
6.
Proc Natl Acad Sci U S A ; 101(24): 9121-6, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15184671

RESUMEN

Significant impairment in endothelial-derived nitric oxide is present in the diabetic corpus cavernosum. RhoA/Rho-kinase may suppress endothelial nitric oxide synthase (eNOS). Here, we tested the hypothesis that RhoA/Rho-kinase contributes to diabetes-related erectile dysfunction and down-regulation of eNOS in the streptozotocin (STZ)-diabetic rat penis. Colocalization of Rho-kinase and eNOS protein was present in the endothelium of the corpus cavernosum. RhoA/Rho-kinase protein abundance and MYPT-1 phosphorylation at Thr-696 were elevated in the STZ-diabetic rat penis. In addition, eNOS protein expression, cavernosal constitutive NOS activity, and cGMP levels were reduced in the STZ-diabetic penis. To assess the functional role of RhoA/Rho-kinase in the penis, we evaluated the effects of an adeno-associated virus encoding the dominant-negative RhoA mutant (AAVTCMV19NRhoA) on RhoA/Rho-kinase and eNOS and erectile function in vivo in the STZ-diabetic rat. STZ-diabetic rats transfected with AAVCMVT19NRhoA had a reduction in RhoA/Rho-kinase and MYPT-1 phosphorylation at a time when cavernosal eNOS protein, constitutive NOS activity, and cGMP levels were restored to levels found in the control rats. There was a significant decrease in erectile response to cavernosal nerve stimulation in the STZ-diabetic rat. AAVT19NRhoA gene transfer improved erectile responses in the STZ-diabetic rat to values similar to control. These data demonstrate a previously undescribed mechanism for the down-regulation of penile eNOS in diabetes mediated by activation of the RhoA/Rho-kinase pathway. Importantly, these data imply that inhibition of RhoA/Rho-kinase improves eNOS protein content and activity thus restoring erectile function in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Disfunción Eréctil/enzimología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Pene/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Adenoviridae/genética , Amidas/farmacología , Animales , Glucemia/metabolismo , Peso Corporal , GMP Cíclico/biosíntesis , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/enzimología , Inhibidores Enzimáticos/metabolismo , Disfunción Eréctil/etiología , Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Masculino , Relajantes Musculares Centrales/farmacología , Fosfatasa de Miosina de Cadena Ligera/biosíntesis , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo III , Pene/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Transfección , Quinasas Asociadas a rho
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA